1
|
Castellano I, Rousset S, Casella D, Capella G, Borella F, Rosa MD, Cassoni P, Catalano A, Ferrante G, Giordano L. Early detection of triple-negative breast cancer: evidence of a favourable prognostic impact in a comparative analysis of screen-detected versus symptomatic cases. BMC Cancer 2025; 25:730. [PMID: 40251506 PMCID: PMC12007119 DOI: 10.1186/s12885-025-14067-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 04/01/2025] [Indexed: 04/20/2025] Open
Abstract
PURPOSE Mammographic screening is effective in reducing breast cancer mortality, but the impact of screening on triple-negative breast cancers (TNBCs) outcomes remains debated. This study aims to determine if screen detection is an independent prognostic factor for TNBCs and to analyse the radiological and pathological differences between screen-detected and symptomatic TNBCs. METHODS This retrospective cohort study analysed 353 histologically confirmed TNBC cases diagnosed between 2013 and 2020 at a single institution in Turin, Italy. Cases were categorized into screen-detected and symptomatic groups based on initial presentation. Clinical, radiological and pathological characteristics as well as disease-free survival (DFS) and overall survival (OS) were compared between groups. Statistical analyses included Kaplan-Meier survival curves and Cox proportional hazard models, adjusting for several clinical and biological variables. RESULTS 50.1% of cases were screen-detected and 49.9% were symptomatic. Screen-detected cases were more commonly smaller (T1 or T2) (96.6%) than symptomatic cases (75%) (p < 0.001). Also, compared to symptomatic tumours, screen-detected ones were more often node negative (62.4% vs. 48%, p = 0.007) and diagnosed at a lower stage (85.4% vs. 63.8%, p < 0.001), with better DFS and OS. Detection method was not an independent prognostic factor, while stage at diagnosis, vascular invasion, histologic type and tumour-infiltrating lymphocytes (TILS) were more significant predictors of prognosis. Radiological and biological features were similar between the two groups. CONCLUSIONS TNBCs correlate with favourable pathological features and improved survival outcomes in univariate analyses, but these benefits diminish when accounting for traditional prognostic factors. Hence, the better prognosis observed among screen-detected cases is more likely due to stage shift rather than tumour biology.
Collapse
Affiliation(s)
| | - Stefano Rousset
- Department of Public Health and Pediatrics, Post Graduate School of Medical Statistics, University of Turin & CPO Piemonte, Turin, Italy.
| | - Denise Casella
- SSD Epidemiologia Screening, CPO-AOU Città Della Salute E Della Scienza Di Torino, Turin, Italy
| | - Giulia Capella
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Fulvio Borella
- Obstetrics and Gynecology Unit 1, Department of Surgical Sciences, Sant'Anna Hospital, University of Turin, Turin, Italy
| | - Martina Di Rosa
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Paola Cassoni
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Alberto Catalano
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
- Centre for Biostatistics, Epidemiology, and Public Health, Department of Clinical and Biological Sciences, University of Turin, Orbassano, TO, Italy
| | - Gianluigi Ferrante
- SSD Epidemiologia Screening, CPO-AOU Città Della Salute E Della Scienza Di Torino, Turin, Italy
| | - Livia Giordano
- SSD Epidemiologia Screening, CPO-AOU Città Della Salute E Della Scienza Di Torino, Turin, Italy
| |
Collapse
|
2
|
Houvenaeghel G, Cohen M, Martino M, Reyal F, Classe JM, Chauvet MP, Colombo PE, Heinemann M, Jouve E, Gimbergues P, Azuar AS, Coutant C, Gonçalves A, de Nonneville A. Negative Survival Impact of Occult Lymph Node Involvement in Small HER2-Positive Early Breast Cancer Treated by Up-Front Surgery. Cancers (Basel) 2023; 15:4567. [PMID: 37760536 PMCID: PMC10526175 DOI: 10.3390/cancers15184567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
(1) Background: The independent negative prognostic value of isolated tumor cells or micro-metastases in axillary lymph nodes has been established in triple-negative breast cancers (BC). However, the prognostic significance of pN0(i+) or pN1mi in HER2-positive BCs treated by primary surgery remains unexplored. Therefore, our objective was to investigate the impact of pN0(i+) or pN1mi in HER2-positive BC patients undergoing up-front surgery on their outcomes. (2) Methods: We retrospectively analyzed 23,650 patients treated in 13 French cancer centers from 1991 to 2013. pN status was categorized as pN0, pN0(i+), pN1mi, and pNmacro. The effect of pN0(i+) or pN1mi on outcomes was investigated both in the entire cohort of patients and in pT1a-b tumors. (3) Results: Of 1771 HER2-positive BC patients included, pN status distributed as follows: 1047 pN0 (59.1%), 60 pN0(i+) (3.4%), 118 pN1mi (6.7%), and 546 pN1 macro-metastases (30.8%). pN status was significantly associated with sentinel lymph node biopsy, axillary lymph node dissection, age, ER status, tumor grade, and size, lymphovascular invasion, adjuvant systemic therapy (ACt), and radiation therapy. With 61 months median follow-up (mean 63.2; CI 95% 61.5-64.9), only pN1 with macro-metastases was independently associated with a negative impact on overall, disease-free, recurrence-free, and metastasis-free survivals in multivariate analysis. In the pT1a-b subgroup including 474 patients, RFS was significantly decreased in multivariate analysis for pT1b BC without ACt (HR 2.365, 1.04-5.36, p = 0.039) and for pN0(i+)/pN1mi patients (HR 2.518, 1.03-6.14, p = 0.042). (4) Conclusions: Survival outcomes were not adversely affected by pN0(i+) and pN1mi in patients with HER2-positive BC. However, in the case of pT1a-b HER2-positive BC, a negative impact on RFS was observed specifically for patients with pN0(i+) and pN1mi diseases, particularly among those with pT1b tumors without ACt. Our findings highlight the importance of considering the pN0(i+) and pN1mi status in the decision-making process when discussing trastuzumab-based ACt for these patients.
Collapse
Affiliation(s)
- Gilles Houvenaeghel
- Department of Surgical Oncology, CRCM, Institut Paoli-Calmettes, Aix-Marseille University, CNRS, INSERM, 232 Bd de Sainte Marguerite, 13009 Marseille, France; (M.C.); (M.M.)
| | - Monique Cohen
- Department of Surgical Oncology, CRCM, Institut Paoli-Calmettes, Aix-Marseille University, CNRS, INSERM, 232 Bd de Sainte Marguerite, 13009 Marseille, France; (M.C.); (M.M.)
| | - Marc Martino
- Department of Surgical Oncology, CRCM, Institut Paoli-Calmettes, Aix-Marseille University, CNRS, INSERM, 232 Bd de Sainte Marguerite, 13009 Marseille, France; (M.C.); (M.M.)
| | - Fabien Reyal
- Institut Curie, 26 Rue d’Ulm, 75248 Paris, France;
| | - Jean-Marc Classe
- Institut René Gauducheau, Site Hospitalier Nord, Boulevard Jacques Monod, 44800 St. Herblain, France;
| | | | | | | | - Eva Jouve
- Centre Claudius Regaud, 20-24 Rue du Pont St. Pierre, 31300 Toulouse, France;
| | - Pierre Gimbergues
- Centre Jean Perrin, 58 Rue Montalembert, 63011 Clermont-Ferrand, France;
| | | | - Charles Coutant
- Centre Georges François Leclerc, 1 Rue du Professeur Marion, 21000 Dijon, France;
| | - Anthony Gonçalves
- Department of Medical Oncology, CRCM, Institut Paoli-Calmettes, Aix-Marseille University, CNRS, INSERM, 232 Bd de Sainte Marguerite, 13009 Marseille, France;
| | - Alexandre de Nonneville
- Department of Medical Oncology, CRCM, Institut Paoli-Calmettes, Aix-Marseille University, CNRS, INSERM, 232 Bd de Sainte Marguerite, 13009 Marseille, France;
| |
Collapse
|
3
|
Null JL, Kim DJ, McCann JV, Pramoonjago P, Fox JW, Zeng J, Kumar P, Edatt L, Pecot CV, Dudley AC. Periostin+ Stromal Cells Guide Lymphovascular Invasion by Cancer Cells. Cancer Res 2023; 83:2105-2122. [PMID: 37205636 PMCID: PMC10330490 DOI: 10.1158/0008-5472.can-22-2412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 02/16/2023] [Accepted: 05/17/2023] [Indexed: 05/21/2023]
Abstract
Cancer cell dissemination to sentinel lymph nodes is associated with poor patient outcomes, particularly in breast cancer. The process by which cancer cells egress from the primary tumor upon interfacing with the lymphatic vasculature is complex and driven by dynamic interactions between cancer cells and stromal cells, including cancer-associated fibroblasts (CAF). The matricellular protein periostin can distinguish CAF subtypes in breast cancer and is associated with increased desmoplasia and disease recurrence in patients. However, as periostin is secreted, periostin-expressing CAFs are difficult to characterize in situ, limiting our understanding of their specific contribution to cancer progression. Here, we used in vivo genetic labeling and ablation to lineage trace periostin+ cells and characterize their functions during tumor growth and metastasis. Periostin-expressing CAFs were spatially found at periductal and perivascular margins, were enriched at lymphatic vessel peripheries, and were differentially activated by highly metastatic cancer cells versus poorly metastatic counterparts. Surprisingly, genetically depleting periostin+ CAFs slightly accelerated primary tumor growth but impaired intratumoral collagen organization and inhibited lymphatic, but not lung, metastases. Periostin ablation in CAFs impaired their ability to deposit aligned collagen matrices and inhibited cancer cell invasion through collagen and across lymphatic endothelial cell monolayers. Thus, highly metastatic cancer cells mobilize periostin-expressing CAFs in the primary tumor site that promote collagen remodeling and collective cell invasion within lymphatic vessels and ultimately to sentinel lymph nodes. SIGNIFICANCE Highly metastatic breast cancer cells activate a population of periostin-expressing CAFs that remodel the extracellular matrix to promote escape of cancer cells into lymphatic vessels and drive colonization of proximal lymph nodes.
Collapse
Affiliation(s)
- Jamie L. Null
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia, Charlottesville, VA 22908, USA
| | - Dae Joong Kim
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia, Charlottesville, VA 22908, USA
| | - James V. McCann
- Department of Cell Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Patcharin Pramoonjago
- Department of Pathology, The University of Virginia, Charlottesville, VA 22908, USA
- UVA Biorepository and Tissue Research Facility
| | - Jay W. Fox
- Emily Couric Comprehensive Cancer Center, The University of Virginia
| | - Jianhao Zeng
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia, Charlottesville, VA 22908, USA
| | - Pankaj Kumar
- UVA Bioinformatics Core
- Department of Biochemistry and Molecular Genetics, The University of Virginia, Charlottesville, VA 22908, USA
| | | | - Chad V. Pecot
- Lineberger Comprehensive Cancer Center
- Division of Hematology/Oncology, Chapel Hill, North Carolina
- UNC RNA Discovery Center
- Department of Medicine, Chapel Hill, North Carolina, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Andrew C. Dudley
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia, Charlottesville, VA 22908, USA
- Emily Couric Comprehensive Cancer Center, The University of Virginia
| |
Collapse
|
4
|
Houvenaeghel G, Cohen M, Classe JM, Reyal F, Mazouni C, Chopin N, Martinez A, Daraï E, Coutant C, Colombo PE, Gimbergues P, Chauvet MP, Azuar AS, Rouzier R, Tunon de Lara C, Muracciole X, Agostini A, Bannier M, Charaffe Jauffret E, De Nonneville A, Goncalves A. Lymphovascular invasion has a significant prognostic impact in patients with early breast cancer, results from a large, national, multicenter, retrospective cohort study. ESMO Open 2021; 6:100316. [PMID: 34864349 PMCID: PMC8645922 DOI: 10.1016/j.esmoop.2021.100316] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/28/2021] [Accepted: 10/31/2021] [Indexed: 11/17/2022] Open
Abstract
Background We determined the prognostic impact of lymphovascular invasion (LVI) in a large, national, multicenter, retrospective cohort of patients with early breast cancer (BC) according to numerous factors. Patients and methods We collected data on 17 322 early BC patients treated in 13 French cancer centers from 1991 to 2013. Survival functions were calculated using the Kaplan–Meier method and multivariate survival analyses were carried out using the Cox proportional hazards regression model adjusted for significant variables associated with LVI or not. Two propensity score-based matching approaches were used to balance differences in known prognostic variables associated with LVI status and to assess the impact of adjuvant chemotherapy (AC) in LVI-positive luminal A-like patients. Results LVI was present in 24.3% (4205) of patients. LVI was significantly and independently associated with all clinical and pathological characteristics analyzed in the entire population and according to endocrine receptor (ER) status except for the time period in binary logistic regression. According to multivariate analyses including ER status, AC, grade, and tumor subtypes, the presence of LVI was significantly associated with a negative prognostic impact on overall (OS), disease-free (DFS), and metastasis-free survival (MFS) in all patients [hazard ratio (HR) = 1.345, HR = 1.312, and HR = 1.415, respectively; P < 0.0001], which was also observed in the propensity score-based analysis in addition to the association of AC with a significant increase in both OS and DFS in LVI-positive luminal A-like patients. LVI did not have a significant impact in either patients with ER-positive grade 3 tumors or those with AC-treated luminal A-like tumors. Conclusion The presence of LVI has an independent negative prognostic impact on OS, DFS, and MFS in early BC patients, except in ER-positive grade 3 tumors and in those with luminal A-like tumors treated with AC. Therefore, LVI may indicate the existence of a subset of luminal A-like patients who may still benefit from adjuvant therapy. In a study of 17 322 early BC patients, LVI had a significant independent negative prognostic impact on survival. LVI negatively impacted survival in almost every patient category and cancer subtype, with and without AC. LVI did not have a negative survival impact in patients with ER+ grade 3 or with luminal A-like tumors with chemotherapy. Results suggest a possible benefit of AC in LVI-positive luminal A-like patients.
Collapse
Affiliation(s)
- G Houvenaeghel
- Department of Surgical Oncology, CRCM, Institut Paoli-Calmettes, Aix-Marseille University, CNRS, INSERM, Marseille, France.
| | - M Cohen
- Department of Surgical Oncology, CRCM, Institut Paoli-Calmettes, Aix-Marseille University, CNRS, INSERM, Marseille, France
| | - J M Classe
- Institut René Gauducheau, Site Hospitalier Nord, St Herblain, France
| | - F Reyal
- Institut Curie, Paris, France
| | - C Mazouni
- Institut Gustave Roussy, Villejuif, France
| | - N Chopin
- Centre Léon Bérard, Lyon, France
| | - A Martinez
- Centre Claudius Regaud, Toulouse, France
| | - E Daraï
- Hôpital Tenon, Paris, France
| | - C Coutant
- Centre Georges François Leclerc, Dijon, France
| | | | | | | | - A S Azuar
- Hôpital de Grasse, Chemin de Clavary, Grasse, France
| | - R Rouzier
- Hôpital René Huguenin, Saint Cloud, France
| | | | | | - A Agostini
- Department of Obstetrics and Gynocology, Hôpital de la Conception, Marseille, France
| | - M Bannier
- Department of Surgical Oncology, CRCM, Institut Paoli-Calmettes, Aix-Marseille University, CNRS, INSERM, Marseille, France
| | - E Charaffe Jauffret
- Department of Pathology, CRCM, Institut Paoli-Calmettes, Aix-Marseille University, Marseille, France
| | - A De Nonneville
- Department of Medical Oncology, CRCM, Institut Paoli-Calmettes, Aix-Marseille University, CNRS, INSERM, Marseille, France
| | - A Goncalves
- Department of Medical Oncology, CRCM, Institut Paoli-Calmettes, Aix-Marseille University, CNRS, INSERM, Marseille, France
| |
Collapse
|
5
|
Hruban RH, Gaida MM, Thompson E, Hong SM, Noë M, Brosens LA, Jongepier M, Offerhaus GJA, Wood LD. Why is pancreatic cancer so deadly? The pathologist's view. J Pathol 2019; 248:131-141. [PMID: 30838636 DOI: 10.1002/path.5260] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/19/2019] [Accepted: 02/28/2019] [Indexed: 12/11/2022]
Abstract
The remarkable aggressiveness of pancreatic cancer has never been fully explained. Although clearly multifactorial, we postulate that venous invasion, a finding seen in most pancreatic cancers but not in most cancers of other organs, may be a significant, underappreciated contributor to the aggressiveness of this disease. Copyright © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Ralph H Hruban
- Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Matthias M Gaida
- Department of General Pathology, The University Hospital of Heidelberg, Heidelberg, Germany
| | - Elizabeth Thompson
- Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Seung-Mo Hong
- Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Michaël Noë
- Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lodewijk Aa Brosens
- Department of Pathology, The University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Martine Jongepier
- Department of Pathology, The University Medical Center Utrecht, Utrecht, The Netherlands
| | - G Johan A Offerhaus
- Department of Pathology, The University Medical Center Utrecht, Utrecht, The Netherlands
| | - Laura D Wood
- Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
6
|
Centonze M, Saponaro C, Mangia A. NHERF1 Between Promises and Hopes: Overview on Cancer and Prospective Openings. Transl Oncol 2018; 11:374-390. [PMID: 29455084 PMCID: PMC5852411 DOI: 10.1016/j.tranon.2018.01.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 01/02/2018] [Accepted: 01/05/2018] [Indexed: 02/07/2023] Open
Abstract
Na+/H+ exchanger regulatory factor 1 (NHERF1) is a scaffold protein, with two tandem PDZ domains and a carboxyl-terminal ezrin-binding (EB) region. This particular sticky structure is responsible for its interaction with different molecules to form multi-complexes that have a pivotal role in a lot of diseases. In particular, its involvement during carcinogenesis and cancer progression has been deeply analyzed in different tumors. The role of NHERF1 is not unique in cancer; its activity is connected to its subcellular localization. The literature data suggest that NHERF1 could be a new prognostic/predictive biomarker from breast cancer to hematological cancers. Furthermore, the high potential of this molecule as therapeutical target in different carcinomas is a new challenge for precision medicine. These evidences are part of a future view to improving patient clinical management, which should allow different tumor phenotypes to be treated with tailored therapies. This article reviews the biology of NHERF1, its engagement in different signal pathways and its involvement in different cancers, with a specific focus on breast cancer. It also considers NHERF1 potential role during inflammation related to most human cancers, designating new perspectives in the study of this "Janus-like" protein.
Collapse
Affiliation(s)
- Matteo Centonze
- Functional Biomorphology Laboratory, IRCCS-Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Concetta Saponaro
- Functional Biomorphology Laboratory, IRCCS-Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Anita Mangia
- Functional Biomorphology Laboratory, IRCCS-Istituto Tumori "Giovanni Paolo II", Bari, Italy.
| |
Collapse
|
7
|
Luo Y, Huang W, Zhang H, Liu G. Prognostic significance of CD117 expression and TP53 missense mutations in triple-negative breast cancer. Oncol Lett 2018; 15:6161-6170. [PMID: 29616097 PMCID: PMC5876428 DOI: 10.3892/ol.2018.8104] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 11/24/2017] [Indexed: 12/15/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is extremely aggressive and associated with poor prognosis. There are no known predictive or prognostic markers for TNBC. Inhibition of tumor protein P53 (TP53) has been demonstrated to increase the levels of cluster of differentiation 117 (CD117) in human colorectal cancer cells. However, the function of TP53 in the regulation of CD117 in TNBC has, to the best of our knowledge, not been reported. In the present study, the association between the expression of CD117 protein and TP53 mutations was investigated, and their prognostic value in patients with TNBC was assessed. A total of 58 TNBC and 48 non-TNBC breast cancer tissue samples were assessed for the expression of CD117, p53 and TP53 mutations. The marker of proliferation Ki-67 (MKI67) proliferation index and vascular invasion index (obtained by measuring D2-40 and CD34) was investigated via immunohistochemistry, and mutations in exons 4–8 of TP53 were measured using direct sequencing. Associations between CD117 and p53 levels or TP53 mutations and clinical parameters were statistically evaluated. The rates of CD117 or MKI67 positivity, CD117+/TP53 missense mutation+, TP53 missense mutations or recurrence were significantly higher in patients with TNBC than in patients with non-TNBC. In TNBC tissues, the presence of CD117 was associated with TP53 missense mutations (P=0.031), vascular invasion, recurrence and MKI67. CD117+/TP53 missense mutation+ also associated with vascular invasion, recurrence and MKI67. Under univariate analysis, MKI67, vascular invasion, CD117, CD117+/TP53 missense mutation+ and TP53 missense mutations were associated with the overall survival of patients with TNBC. Multivariate analysis revealed that vascular invasion and CD117+/TP53 missense mutation+ in primary tumors were independent prognostic factors in patients with TNBC. In conclusion, CD117+/TP53 missense mutation+ was associated with MKI67, vascular invasion and tumor recurrence in TNBC. The presence of CD117 and TP53 missense mutations together in the primary tumors was an independent prognostic factor for survival of patients with TNBC.
Collapse
Affiliation(s)
- Yanli Luo
- Department of Pathology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Wentao Huang
- Department of Pathology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Huizhen Zhang
- Department of Pathology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Guang Liu
- Department of Vascular Surgery, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, P.R. China
| |
Collapse
|
8
|
Xu T, Zhang L, Xu H, Kang S, Xu Y, Luo X, Hua T, Tang G. Prediction of low-risk breast cancer using quantitative DCE-MRI and its pathological basis. Oncotarget 2017; 8:114360-114370. [PMID: 29371992 PMCID: PMC5768409 DOI: 10.18632/oncotarget.22267] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 07/26/2017] [Indexed: 12/17/2022] Open
Abstract
Purpose This study aimed to evaluate the difference of mass in dynamic contrast enhanced magnetic resonance imaging (DCE-MRI) characteristics between low-risk and non-low-risk breast cancers and to explore the possible pathological basis. Materials and Methods Approval from the institutional review board and informed consent were acquired for this study. The MR images of 104 patients with pathologically proven breast cancer (104 lesions) were prospectively analyzed. All of included patients were Chinese woman. The DCE-MRI morphologic findings, apparent diffusion coefficient (ADC) values, quantitative DCE-MRI parameters, and pathological biomarkers between the two subtypes of breast cancer were compared. The quantitative DCE-MRI parameters and ADC values were added to the morphologic features in multivariate models to evaluate diagnostic performance in predicting low-risk breast cancer. The values were further subjected to the receiver operating characteristic (ROC) curve analysis. Results Low-risk tumors showed significantly lower Ktrans and Kepvalues (t = 2.065, P = 0.043 and t = 3.548, P = 0.001, respectively) and higher ADC value (t = 4.713, P = 0.000) than non-low-risk breast cancers. Our results revealed no significant differences in clinic data and conventional imaging findings between the two breast cancer subtypes. Adding the quantitative DCE-MRI parameters and ADC values to conventional MRI improved the diagnostic performance of MRI: The area under the ROC improved from 0.63 to 0.91. Low-risk breast cancers showed significantly lower matrix metalloproteinase (MMP)-2 expression (P = 0.000), lower MMP-9 expression (P = 0.001), and lower microvessel density (MVD) values (P = 0.008) compared with non-low-risk breast cancers. Ktrans and Kep values were positively correlated with pathological biomarkers. The ADC value showed a significant inverse correlation with pathological biomarkers. Conclusions The prediction parameter using Ktrans, Kep, and ADC obtained on DCE-MRI and diffusion-weighted imaging could facilitate the identification of low-risk breast cancers. Decreased biological factors, including MVD, vascular endothelial growth factor, MMP-2, and MMP-9, may explain the possible pathological basis.
Collapse
Affiliation(s)
- Tingting Xu
- Department of Radiology, Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Lin Zhang
- Department of Radiology, Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Hong Xu
- Department of Radiology, Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Sifeng Kang
- Department of Radiology, Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Yali Xu
- Department of Radiology, Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Xiaoyu Luo
- Department of Radiology, Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Ting Hua
- Department of Radiology, Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Guangyu Tang
- Department of Radiology, Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| |
Collapse
|
9
|
de Nonneville A, Gonçalves A, Zemmour C, Cohen M, Classe JM, Reyal F, Colombo PE, Jouve E, Giard S, Barranger E, Sabatier R, Bertucci F, Boher JM, Houvenaeghel G. Adjuvant chemotherapy in pT1ab node-negative triple-negative breast carcinomas: Results of a national multi-institutional retrospective study. Eur J Cancer 2017; 84:34-43. [PMID: 28780480 DOI: 10.1016/j.ejca.2017.06.043] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 06/19/2017] [Accepted: 06/27/2017] [Indexed: 12/27/2022]
Abstract
BACKGROUND Triple-negative breast cancers (TNBCs) are considered as associated with poor outcome, but prognosis of subcentimetric, node-negative disease remains controversial and evidence that adjuvant chemotherapy (CT) is effective in these small tumours remains limited. PATIENTS AND METHODS Our objective was to investigate the impact of CT on survival in pT1abN0M0 TNBC. Patients were retrospectively identified from a cohort of 22,475 patients who underwent primary surgery in 15 French centres between 1987 and 2013. As rare pathological types may display very particular prognoses in these tumours, we retained only the invasive ductal carcinomas of no special type according to the last World Health Organisation (WHO) classification which is the most common TNBC histological type. End-points were disease-free survival (DFS) and metastasis-free survival (MFS). A propensity score for receiving CT was estimated using a logistic regression including age, tumour size, Scarff Bloom and Richardson (SBR) grade and lymphovascular invasion. RESULTS Of a total of 284 patients with pT1abN0M0 ductal TNBC, 144 (51%) received CT and 140 (49%) did not. Patients receiving CT had more adverse prognostic features, such as tumour size, high grade, young age, and lymphovascular invasion. CT was not associated with a significant benefit for DFS (Hazard ratio, HR = 0.77 [0.40-1.46]; p = 0.419, log-rank test) or MFS (HR = 1.00 [0.46-2.19]; p = 0.997), with 5-year DFS and MFS in the group with CT versus without of 90% [81-94%] versus 84% [74-90%], and 90% [81-95%] versus 90% [83%-95%], respectively. Results were consistent in all supportive analyses including multivariate Cox model and the use of the propensity score for adjustment and as a matching factor for case-control analyses. CONCLUSIONS This study did not identify a significant DFS or MFS advantage for CT in subcentimetric, node-negative ductal TNBC. Although current consensus guidelines recommend consideration of CT in all TNBC larger than 5 mm, clinicians should carefully discuss benefit/risk ratio with patients, given the unproven benefits.
Collapse
Affiliation(s)
- A de Nonneville
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France.
| | - A Gonçalves
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France.
| | - C Zemmour
- Department of Clinical Research and Investigation, Biostatistics and Methodology Unit, Institut Paoli-Calmettes, Aix Marseille University, INSERM, IRD, SESSTIM, Marseille, France
| | - M Cohen
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Department of Surgical Oncology, CRCM, Marseille, France
| | - J M Classe
- Institut René Gauducheau, Saint-Herblain, France
| | - F Reyal
- Institut Curie, Paris, France
| | | | - E Jouve
- Institut Claudius Regaud, Toulouse, France
| | - S Giard
- Centre Oscar Lambret, Lille, France
| | | | - R Sabatier
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - F Bertucci
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - J M Boher
- Department of Clinical Research and Investigation, Biostatistics and Methodology Unit, Institut Paoli-Calmettes, Aix Marseille University, INSERM, IRD, SESSTIM, Marseille, France
| | - G Houvenaeghel
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Department of Surgical Oncology, CRCM, Marseille, France
| |
Collapse
|
10
|
Xue S, He L, Zhang X, Zhou J, Li F, Wang X. Expression of Jagged1/Notch3 Signaling Pathway and their Relationship with the Tumor Angiogenesis in TNBC. Arch Med Res 2017. [PMID: 28625320 DOI: 10.1016/j.arcmed.2017.03.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Jagged1/Notch3 signaling pathway plays a key role in angiogenesis of breast cancer, but little is known in TNBC. This study was designed to investigate the expression of Jagged1/Notch3 mRNA and protein in TNBC, analyze their correlations with clinicopathological characteristics and prognosis. Moreover, the interrelationship among Jagged1/Notch3 and VEGF was initially evaluated. METHODS Jagged1/Notch3 mRNA and protein expression levels were determined by Q-RT-PCR and Western blotting. Additionally, Immunohistochemistry for Jagged1/Notch3 was detected by Ventana platform, VEGF and CD34 was performed using the EnVision/HRP technique. RESULTS mRNA transcriptionof Jagged1/Notch3 was in accord with protein expression. TNBC patients with positive Jagged1 expression had poorer DFS (p = 0.008) and OS (p = 0.004). Jagged1 expression was independent predictors of OS (p = 0.038). The expression of VEGF was positively correlative to MVD (p = 0.018), MVD was significantly associated with Jagged1 (p <0.0001) and Notch3 (p <0.0001). The expression of Jagged1/Notch3 has no correlation with VEGF, only in positive VEGF expression of TNBC patients Jagged1/Notch3 had influence on DFS and OS (p <0.05). CONCLUSION Jagged1/Notch3 was -expressed at both the mRNA and protein levels, Jagged1 served as an independent predictor of poor prognosis. We speculate that there is a cross-talk between Jagged1/Notch3 and VEGF in TNBC angiogenesis. Jagged1/Notch3 is expected to be an important signaling pathway for TNBC progression and a potential target for TNBC neovascularization therapy.
Collapse
Affiliation(s)
- Siliang Xue
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lang He
- Cancer Center, The Second Clinical Medical College of North Sichuan Medical College, Nanchong Central Hospital, Nanchong, Sichuan, China
| | - Xiao Zhang
- Department of Breast Surgery, Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Jin Zhou
- Department of Chemotherapy, Sichuan Cancer Hospital, Chengdu, Sichuan, China
| | - Fanghua Li
- Department of Pathology, Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Xiaoshan Wang
- Cancer Center, Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China.
| |
Collapse
|
11
|
Hwang KT, Kim YA, Kim J, Chu AJ, Chang JH, Oh SW, Hwang KR, Chai YJ. The influences of peritumoral lymphatic invasion and vascular invasion on the survival and recurrence according to the molecular subtypes of breast cancer. Breast Cancer Res Treat 2017; 163:71-82. [PMID: 28194538 DOI: 10.1007/s10549-017-4153-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 02/07/2017] [Indexed: 01/02/2023]
Abstract
PURPOSE We aimed to compare the influences of lymphatic invasion (LI) and vascular invasion (VI) on survival and recurrence according to the molecular subtypes of breast cancer. METHODS We retrospectively analyzed data on 820 breast cancer patients and assessed overall survival (OS) and disease-free survival (DFS) according to LI and VI using the Kaplan-Meier estimator and the Cox proportional hazards model. RESULTS Both positive LI and positive VI showed inferior OS and DFS compared with negative LI and negative VI (all p < 0.001). Both positive LI and positive VI showed higher local, regional, and distant recurrence rates (p = 0.002 for regional recurrence of VI, p < 0.001 for all the others). Although LI was a significant independent predictor of OS (hazard ratio [HR] 1.927; 95% confidence interval [CI] 1.046-3.553) and DFS (HR 1.815; 95% CI 1.063-3.096), VI was not in the multivariate analyses. Regarding OS, both positive LI and positive VI showed worse survival rates in the luminal A (p = 0.016 and p = 0.024, respectively) and triple negative subtypes (both p < 0.001). Regarding DFS, LI was a significant prognosticator in the luminal A and triple negative (both p < 0.001) subtypes. VI was a significant prognosticator across all molecular subtypes, although the prognostic impact was most prominent in the luminal A subtype (p < 0.001). CONCLUSIONS Both LI and VI were significant, unfavorable prognostic factors of OS and DFS, especially in the luminal A and triple negative breast cancer subtypes. Although LI was a significant independent predictor of OS and DFS, VI was not after the multivariate analyses.
Collapse
Affiliation(s)
- Ki-Tae Hwang
- Department of Surgery, Seoul National University Boramae Medical Center, 39, Boramae-Gil, Dongjak-gu, Seoul, 156-707, Republic of Korea.
| | - Young A Kim
- Department of Pathology, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Jongjin Kim
- Department of Surgery, Seoul National University Boramae Medical Center, 39, Boramae-Gil, Dongjak-gu, Seoul, 156-707, Republic of Korea
| | - A Jung Chu
- Department of Radiology, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Ji Hyun Chang
- Department of Radiation Oncology, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - So Won Oh
- Department of Nuclear Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Kyu Ri Hwang
- Department of Obstetrics and Gynecology, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Young Jun Chai
- Department of Surgery, Seoul National University Boramae Medical Center, 39, Boramae-Gil, Dongjak-gu, Seoul, 156-707, Republic of Korea
| |
Collapse
|
12
|
Houvenaeghel G, Sabatier R, Reyal F, Classe JM, Giard S, Charitansky H, Rouzier R, Faure C, Garbay JR, Daraï E, Hudry D, Gimbergues P, Villet R, Lambaudie E. Axillary lymph node micrometastases decrease triple-negative early breast cancer survival. Br J Cancer 2016; 115:1024-1031. [PMID: 27685443 PMCID: PMC5117781 DOI: 10.1038/bjc.2016.283] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 07/19/2016] [Accepted: 08/09/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Triple-negative breast cancers (TNBCs) are the most deadly form of breast cancer (BC) subtypes. Axillary lymph node involvement (ALNI) has been described to be prognostic in BC taken as a whole, but its prognostic value in each subtype is unclear. We explored the prognostic impact of ALNI and especially of small size axillary metastases in early TNBCs. METHODS We analysed in this multicentre study all patients treated for early TNBC in 12 French cancer centres. We explored the correlation between clinicopathological data and ALNI, with a specific focus on the dichotomisation between macrometastases and occult metastases, which is defined as the presence of isolated tumour cells or micrometastases. The prognostic value of ALNI both in terms of disease-free survival (DFS) and overall survival (OS) was also explored. RESULTS We included 1237 TNBC patients. Five-year DFS and OS were 83.7% and 88.5%, respectively. The identified independent prognostic features for DFS were tumour size >20 mm (hazard ratio (HR)=1.86; 95% CI: 1.11-3.10, P=0.018), lymphovascular invasion (HR=1.69; 95% CI: 1.21-2.34, P=0.002) and ALNI both in case of macrometastases (HR=1.97; 95% CI: 1.38-2.81, P<0.0001) and occult metastases (HR=1.72; 95% CI: 1.1-2.71, P=0.019). DFS and OS were similar between tumours with occult metastases and macrometastases. Tumours presenting at least two pejorative features (out of ALNI, lymphovascular invasion and large tumour size) displayed a significantly poorer DFS in both the training set and validation set, independently of chemotherapy administration. Tumours with no more than one of the above-cited pejorative features had a 5-year OS of ⩾90% vs 70% for other cases (P<0.0001). CONCLUSIONS Axillary lymph node involvement is a key prognostic feature for early TNBC when isolated tumour cells were identified in lymph nodes. This impact is independent of chemotherapy use.
Collapse
Affiliation(s)
- G Houvenaeghel
- Institut Paoli Calmettes and Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS U7258, 232 Bd Ste Marguerite, Marseille, France
| | - R Sabatier
- Institut Paoli Calmettes and Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS U7258, 232 Bd Ste Marguerite, Marseille, France
| | - F Reyal
- Institut Curie, Paris, France
| | - J M Classe
- Institut René Gauducheau, Site hospitalier Nord, St Herblain, France
| | - S Giard
- Centre Oscar Lambret, 3 rue Frédéric Combenal, Lille, France
| | - H Charitansky
- Centre Claudius Regaud, 20-24 rue du Pont St Pierre, Toulouse, France
| | - R Rouzier
- Centre René Huguenin, 35 rue Dailly, Saint Cloud, France
| | - C Faure
- Centre Léon Bérard, 28 rue Laennec, Lyon, France
| | - J R Garbay
- Institut Gustave Roussy, 114 rue Edouard Vaillant, Villejuif, France
| | - E Daraï
- Hôpital Tenon, 4 rue de la Chine, Paris, France
| | - D Hudry
- Centre Georges François Leclerc, 1 rue du Professeur Marion, Dijon, France
| | - P Gimbergues
- Centre Jean Perrin, 58 rue Montalembert, Clermont Ferrand, France
| | - R Villet
- Hôpital des Diaconnesses, 18 rue du Sergent Bauchat, Paris, France
| | - E Lambaudie
- Institut Paoli Calmettes and Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS U7258, 232 Bd Ste Marguerite, Marseille, France
| |
Collapse
|
13
|
Rahal S, Boher JM, Extra JM, Tarpin C, Charafe-Jauffret E, Lambaudie E, Sabatier R, Thomassin-Piana J, Tallet A, Resbeut M, Houvenaeghel G, Laborde L, Bertucci F, Viens P, Gonçalves A. Immunohistochemical subtypes predict the clinical outcome in high-risk node-negative breast cancer patients treated with adjuvant FEC regimen: results of a single-center retrospective study. BMC Cancer 2015; 15:697. [PMID: 26466893 PMCID: PMC4607139 DOI: 10.1186/s12885-015-1746-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 10/09/2015] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Anthracycline-based adjuvant chemotherapy improves survival in patients with high-risk node-negative breast cancer (BC). In this setting, prognostic factors predicting for treatment failure might help selecting among the different available cytotoxic combinations. METHODS Between 1998 and 2008, 757 consecutive patients with node-negative BC treated in our institution with adjuvant FEC (5FU, epirubicin, cyclophosphamide) chemotherapy were identified. Data collection included demographic, clinico-pathological characteristics and treatment information. Molecular subtypes were derived from estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2) status and Scarff-Bloom-Richardson (SBR) grade. Disease-free survival (DFS), distant disease-free survival (DDFS) and overall survival (OS) were estimated using the Kaplan-Meier Method, and prognostic factors were examined by multivariate Cox analysis. RESULTS After a median follow-up of 70 months, the 5-year DFS, DDFS and OS were 90.6 % (95 % confidence interval (CI): 88.2-93.1), 92.8 % (95 % CI: 90.7-95) and 95.1 % (95 % CI, 93.3-96.9), respectively. In the multivariate analysis including classical clinico-pathological parameters, only grade 3 maintained a significant and independent adverse prognostic impact. In an alternative multivariate model where ER, PR and grade were replaced by molecular subtypes, only luminal B/HER2-negative and triple-negative subtypes were associated with reduced DFS and DDFS. CONCLUSIONS Node-negative BC patients receiving adjuvant FEC regimen have a favorable outcome. Luminal B/HER2-negative and triple-negative subtypes identify patients with a higher risk of treatment failure, which might warrant more aggressive systemic treatment.
Collapse
Affiliation(s)
- S Rahal
- Department of Medical Oncology, Institut Paoli-Calmettes, 232 Bd. Sainte-Marguerite, 13009, Marseille, France.
| | - J M Boher
- Department of Biostatistics, Institut Paoli-Calmettes, Marseille, France.
- Centre de Recherche en Cancérologie de Marseille, U1068 INSERM, U7258 CNRS, Marseille, France.
| | - J M Extra
- Department of Medical Oncology, Institut Paoli-Calmettes, 232 Bd. Sainte-Marguerite, 13009, Marseille, France.
- Centre de Recherche en Cancérologie de Marseille, U1068 INSERM, U7258 CNRS, Marseille, France.
| | - C Tarpin
- Department of Medical Oncology, Institut Paoli-Calmettes, 232 Bd. Sainte-Marguerite, 13009, Marseille, France.
| | - E Charafe-Jauffret
- Department of Biopathology, Institut Paoli-Calmettes, Marseille, France.
- Centre de Recherche en Cancérologie de Marseille, U1068 INSERM, U7258 CNRS, Marseille, France.
- Aix-Marseille University, Marseille, France.
| | - E Lambaudie
- Department of Surgical Oncology, Institut Paoli-Calmettes, Marseille, France.
- Centre de Recherche en Cancérologie de Marseille, U1068 INSERM, U7258 CNRS, Marseille, France.
| | - R Sabatier
- Department of Medical Oncology, Institut Paoli-Calmettes, 232 Bd. Sainte-Marguerite, 13009, Marseille, France.
- Centre de Recherche en Cancérologie de Marseille, U1068 INSERM, U7258 CNRS, Marseille, France.
- Aix-Marseille University, Marseille, France.
| | - J Thomassin-Piana
- Department of Biopathology, Institut Paoli-Calmettes, Marseille, France.
- Centre de Recherche en Cancérologie de Marseille, U1068 INSERM, U7258 CNRS, Marseille, France.
| | - A Tallet
- Department of Radiation Oncology, Institut Paoli-Calmettes, Marseille, France.
| | - M Resbeut
- Department of Radiation Oncology, Institut Paoli-Calmettes, Marseille, France.
| | - G Houvenaeghel
- Department of Surgical Oncology, Institut Paoli-Calmettes, Marseille, France.
- Centre de Recherche en Cancérologie de Marseille, U1068 INSERM, U7258 CNRS, Marseille, France.
- Aix-Marseille University, Marseille, France.
| | - L Laborde
- Data Management and Analysis Center, Institut Paoli-Calmettes, Marseille, France.
| | - F Bertucci
- Department of Medical Oncology, Institut Paoli-Calmettes, 232 Bd. Sainte-Marguerite, 13009, Marseille, France.
- Centre de Recherche en Cancérologie de Marseille, U1068 INSERM, U7258 CNRS, Marseille, France.
- Aix-Marseille University, Marseille, France.
| | - P Viens
- Department of Medical Oncology, Institut Paoli-Calmettes, 232 Bd. Sainte-Marguerite, 13009, Marseille, France.
- Centre de Recherche en Cancérologie de Marseille, U1068 INSERM, U7258 CNRS, Marseille, France.
- Aix-Marseille University, Marseille, France.
| | - A Gonçalves
- Department of Medical Oncology, Institut Paoli-Calmettes, 232 Bd. Sainte-Marguerite, 13009, Marseille, France.
- Centre de Recherche en Cancérologie de Marseille, U1068 INSERM, U7258 CNRS, Marseille, France.
- Aix-Marseille University, Marseille, France.
| |
Collapse
|
14
|
Sugiyama M, Hasebe T, Shimada H, Takeuchi H, Shimizu K, Shimizu M, Yasuda M, Ueda S, Shigekawa T, Osaki A, Saeki T. Grading system for blood vessel tumor emboli of invasive ductal carcinoma of the breast. Hum Pathol 2015; 46:906-16. [DOI: 10.1016/j.humpath.2015.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 03/01/2015] [Accepted: 03/06/2015] [Indexed: 12/12/2022]
|
15
|
Gujam FJ, Going JJ, Edwards J, Mohammed ZM, McMillan DC. The role of lymphatic and blood vessel invasion in predicting survival and methods of detection in patients with primary operable breast cancer. Crit Rev Oncol Hematol 2014; 89:231-41. [DOI: 10.1016/j.critrevonc.2013.08.014] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 08/21/2013] [Accepted: 08/30/2013] [Indexed: 01/03/2023] Open
|
16
|
Kim KJ, Godarova A, Seedle K, Kim MH, Ince TA, Wells SI, Driscoll JJ, Godar S. Rb suppresses collective invasion, circulation and metastasis of breast cancer cells in CD44-dependent manner. PLoS One 2013; 8:e80590. [PMID: 24324613 PMCID: PMC3851742 DOI: 10.1371/journal.pone.0080590] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 10/04/2013] [Indexed: 01/06/2023] Open
Abstract
Basal-like breast carcinomas (BLCs) present with extratumoral lymphovascular invasion, are highly metastatic, presumably through a hematogenous route, have augmented expression of CD44 oncoprotein and relatively low levels of retinoblastoma (Rb) tumor suppressor. However, the causal relation among these features is not clear. Here, we show that Rb acts as a key suppressor of multiple stages of metastatic progression. Firstly, Rb suppresses collective cell migration (CCM) and CD44-dependent formation of F-actin positive protrusions in vitro and cell-cluster based lymphovascular invasion in vivo. Secondly, Rb inhibits the release of single cancer cells and cell clusters into the hematogenous circulation and subsequent metastatic growth in lungs. Finally, CD44 expression is required for collective motility and all subsequent stages of metastatic progression initiated by loss of Rb function. Altogether, our results suggest that Rb/CD44 pathway is a crucial regulator of CCM and metastatic progression of BLCs and a promising target for anti-BLCs therapy.
Collapse
MESH Headings
- Actins/genetics
- Actins/metabolism
- Animals
- Carcinoma, Basal Cell/genetics
- Carcinoma, Basal Cell/metabolism
- Carcinoma, Basal Cell/secondary
- Cell Line, Tumor
- Cell Movement
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Hyaluronan Receptors/genetics
- Hyaluronan Receptors/metabolism
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/secondary
- Lymphatic Metastasis
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Mice
- Neoplastic Cells, Circulating/metabolism
- Neoplastic Cells, Circulating/pathology
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Retinoblastoma Protein/antagonists & inhibitors
- Retinoblastoma Protein/genetics
- Retinoblastoma Protein/metabolism
Collapse
Affiliation(s)
- Kui-Jin Kim
- Department of Cancer Biology, Vontz Center for Molecular Studies, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Alzbeta Godarova
- Department of Cancer Biology, Vontz Center for Molecular Studies, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Kari Seedle
- Department of Cancer Biology, Vontz Center for Molecular Studies, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Min-Ho Kim
- Biomedical Research Center, Ulsan University Hospital, Ulsan, Republic of Korea
| | - Tan A. Ince
- Department of Pathology, Braman Family Breast Cancer Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Susanne I. Wells
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - James J. Driscoll
- Department of Cancer Biology, Vontz Center for Molecular Studies, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- Division of Hematology and Oncology, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Samuel Godar
- Department of Cancer Biology, Vontz Center for Molecular Studies, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- * E-mail:
| |
Collapse
|
17
|
Dekker TJA, van de Velde CJH, van Bruggen D, Mesker WE, van der Hoeven JJM, Kroep JR, Tollenaar RAEM, Smit VTHBM. Quantitative assessment of lymph vascular space invasion (LVSI) provides important prognostic information in node-negative breast cancer. Ann Oncol 2013; 24:2994-8. [PMID: 24114856 DOI: 10.1093/annonc/mdt400] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Some studies investigating the prognostic value of lymph vascular space invasion (LVSI) have shown an association between LVSI and disease-free survival. Definitive criteria and optimal determination of this parameter remain unclear, however, especially regarding the clinical relevance of LVSI quantification. PATIENTS AND METHODS A subset of node-negative breast carcinomas from premenopausal patients from the European Organization for the Research and Treatment of Cancer trial 10854 (assessing efficacy of perioperative chemotherapy patients with T1-T3, N0-2, and M0 breast cancer (BC) was selected and scored for LVSI. In 358 evaluable breast carcinomas, the number of LVSI foci and tumor cells was determined in the largest tumor embolus within the lymph vessels. These two parameters were multiplied to calculate the LVSI tumor burden (LVSI TB). The optimal cutoff for this parameter was calculated in a test set (N = 120), tested in a validation set (N = 238), and compared with simple quantitation of the number of LVSI foci. RESULTS Tumors with a single LVSI focus are not associated with increased risk for relapse [hazard ratio (HR) 1.423, 95% confidence interval (CI) 0.762-2.656]. The LVSI TB had higher sensitivity and specificity compared with simple determination of the number of LVSI foci. LVSI TB was independently associated with disease-free survival in the validation set (HR 2.366, 95% CI 1.369-4.090, P = 0.002) in multivariate analysis and provided prognostic information in both the low- and high-risk node-negative BC groups (P < 0.001 and P = 0.007, respectively). CONCLUSION The determination of the number of LVSI foci multiplied by the number of tumor cells gives the most reliable quantitative assessment of this parameter, which can provide prognostic information in node-negative BC.
Collapse
|
18
|
[Triple-negative breast cancer: histoclinical and molecular features, therapeutic management and perspectives]. Bull Cancer 2013; 100:453-64. [PMID: 23695030 DOI: 10.1684/bdc.2013.1740] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Triple-negative breast cancer (TNBC), as defined by the absence of estrogen and progesterone receptor expression, as well as the lack of HER2 overexpression/amplification, corresponds to 15% of breast cancer and represents an aggressive form of the disease. TNBC are frequently confounded with basal subtype in the molecular classification of breast cancer and also share some similarities with BRCA1-mutated tumors. Epidemiological and clinical characteristics are distinct from other subtypes, including a younger age at diagnosis, a higher risk of relapse in spite of increased chemosensitivity, and a higher incidence of lung and brain metastatic relapses. Conventional cytotoxics remain the mainstay of current systemic management but recent evaluation of more targeted therapeutics, including specific cytotoxics (such as the use of platinum salts), PARP and EGFR inhibition, and antiangiogenics have been performed, providing contrasted but rather disappointing results. Recent data indicate that TNBC represent a heterogeneous entity composed of multiple and distinct molecular subtypes, which should deserve specific targeted therapeutics.
Collapse
|
19
|
Novel model for basaloid triple-negative breast cancer: behavior in vivo and response to therapy. Neoplasia 2013; 14:926-42. [PMID: 23097627 DOI: 10.1593/neo.12956] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 08/16/2012] [Accepted: 08/17/2012] [Indexed: 12/25/2022] Open
Abstract
INTRODUCTION The basaloid triple-negative breast cancer (B-TNBC) is one of the most aggressive, therapy-resistant, and metastatic tumors. Current models do not recapitulate the basaloid phenotype of TNBC, thus limiting the understanding of its biology and designing new treatments. We identified HCC1806 as a line expressing typical B-TNBC markers, engineered a subline with traceable reporters, and determined growth, drug sensitivity, recurrence, and vascular and metastatic patterns of orthotopic xenografts in immunodeficient mice. METHODS mRNA and protein analyses showed that HCC1806 expresses basal but not luminal or mesenchymal markers. HCC1806-RR subline stably expressing red fluorescent protein and Renilla luciferase was generated and characterized for sensitivity to chemodrugs, orthotopic growth, vascular properties, recurrence, metastasis, and responsiveness in vivo. RESULTS The HCC1806 cells were highly sensitive to paclitaxel, but cytotoxicity was accompanied by pro-survival vascular endothelial growth factor-A loop. In vivo, HCC1806-RR tumors display linear growth, induce peritumoral lymphatics, and spontaneously metastasize to lymph nodes (LNs) and lungs. Similarly to human B-TNBC, HCC1806-RR tumors were initially sensitive to taxane therapy but subsequently recur. Bevacizumab significantly suppressed recurrence by 50% and reduced the incidence of LN and pulmonary metastases by, respectively, 50% and 87%. CONCLUSIONS The HCC1806-RR is a new model that expresses bona fide markers of B-TNBC and traceable markers for quantifying metastases. Combination of bevacizumab with nab-paclitaxel significantly improved the outcome, suggesting that this approach can apply to human patients with B-TNBC. This model can be used for defining the metastatic mechanisms of B-TNBC and testing new therapies.
Collapse
|
20
|
Synnestvedt M, Borgen E, Russnes HG, Kumar NT, Schlichting E, Giercksky KE, Kåresen R, Nesland JM, Naume B. Combined analysis of vascular invasion, grade, HER2 and Ki67 expression identifies early breast cancer patients with questionable benefit of systemic adjuvant therapy. Acta Oncol 2013; 52:91-101. [PMID: 22934555 DOI: 10.3109/0284186x.2012.713508] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Over-treatment of low-risk early breast cancer patients with adjuvant systemic therapies is an important clinical challenge. Better techniques are required which can be used to distinguish between the large group of patients with no residual disease after surgery and consequently no benefit of adjuvant treatment, from the smaller group with high relapse risk. A better integration of available prognostic factors might contribute to improved prediction of clinical outcome. MATERIAL AND METHODS The current study included 346 unselected pT1pN0 patients who did not receive adjuvant systemic treatment. In Norway, no patients with this stage were recommended systemic treatment at the time of the study (1995-1998). Histological type, tumour size, grade, vascular invasion (VI), hormone receptor (HR) status, HER2 and Ki67 (cut-off 10%) were analysed. Median follow-up was 86 months for relapse and 101 months for death. RESULTS Thirty-eight patients experienced relapse, 31 with distant metastasis. Twenty-one patients died of breast cancer. In univariate analysis grade, HER2, HR, VI and Ki67 had impact on clinical outcome (p < 0.005, log rank). In multivariate analysis, only grade 1-2 vs. grade 3, HER2, VI, and Ki67 status were significant for disease free survival, distant disease free survival, and/or breast cancer specific survival. These factors were used in combination, to separate patients into groups based on the number of unfavourable factors present [combined prognostic score (CPS) 0-4]. Close to 2/3 of the patients (61.4%) had no unfavourable factor (CPS0), whilst 18.4% had CPS ≥ 2. Only 3.6% of those with CPS0 developed metastasis (p < 0.001). The outcome was clearly worse for patients with CPS ≥ 2 (p < 0.001), systemic relapse was detected in approximately 40%. CONCLUSIONS This study indicates that the combined use of grade, VI, HER2 and Ki67 identifies a subgroup of breast cancer patients with a relapse risk that may question the benefit of adjuvant systemic therapy.
Collapse
Affiliation(s)
- Marit Synnestvedt
- Department of Oncology, Division of Surgery and Cancer Medicine, Oslo University Hospital, Radiumhospitalet, Oslo, Norway
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Bordbar E, Malekzadeh M, Ardekani MTF, Doroudchi M, Ghaderi A. Serum Levels of G-CSF and IL-7 in Iranian Breast Cancer Patients. Asian Pac J Cancer Prev 2012; 13:5307-12. [DOI: 10.7314/apjcp.2012.13.10.5307] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
22
|
Malfettone A, Saponaro C, Paradiso A, Simone G, Mangia A. Peritumoral vascular invasion and NHERF1 expression define an immunophenotype of grade 2 invasive breast cancer associated with poor prognosis. BMC Cancer 2012; 12:106. [PMID: 22439624 PMCID: PMC3362775 DOI: 10.1186/1471-2407-12-106] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Accepted: 03/22/2012] [Indexed: 11/10/2022] Open
Abstract
Background Traditional determinants proven to be of prognostic importance in breast cancer include the TNM staging, histological grade, proliferative activity, hormone receptor status and HER2 overexpression. One of the limitations of the histological grading scheme is that a high percentage of breast cancers are still classified as grade 2, a category with ambiguous clinical significance. The aim of this study was to best characterize tumors scored as grade 2. Methods We investigated traditional prognostic factors and a panel of tumor markers not used in routine diagnosis, such as NHERF1, VEGFR1, HIF-1α and TWIST1, in 187 primary invasive breast cancers by immunohistochemistry, stratifying patients into good and poor prognostic groups by the Nottingham Prognostic Index. Results Grade 2 subgroup analysis showed that the PVI (p = 0.023) and the loss of membranous NHERF1 (p = 0.028) were adverse prognostic factors. Relevantly, 72% of grade 2 tumors were associated to PVI+/membranous NHERF1- expression phenotype, characterizing an adverse prognosis (p = 0.000). Multivariate logistic regression analysis in the whole series revealed poor prognosis correlated with PVI and MIB1 (p = 0.000 and p = 0.001, respectively). Furthermore, in the whole series of breast cancers we found cytoplasmic NHERF1 expression positively correlated to VEGFR1 (r = 0.382, p = 0.000), and in VEGFR1-overexpressing tumors the oncogenic receptor co-localized with NHERF1 at cytoplasmic level. Conclusions The PVI+/membranous NHERF1- expression phenotype identifies a category of grade 2 tumors with the worst prognosis, including patient subgroup with a family history of breast cancer. These observations support the idea of the PVI+/membranous NHERF1- expression immunophenotype as a useful marker, which could improve the accuracy of predicting clinical outcome in grade 2 tumors.
Collapse
Affiliation(s)
- Andrea Malfettone
- Functional Biomorphology Laboratory, National Cancer Centre, Bari, Italy
| | | | | | | | | |
Collapse
|
23
|
Rubovszky G, Horváth Z, Tóth E, Láng I, Kásler M. Significance of histomorphology of early triple-negative breast cancer. Pathol Oncol Res 2012; 18:823-31. [PMID: 22415664 DOI: 10.1007/s12253-012-9510-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Accepted: 02/20/2012] [Indexed: 01/03/2023]
Abstract
UNLABELLED Triple-negative breast cancer (TNBC) is a heterogeneous disease. Possibly genetic characterisation provides the most appropriate information on tumour biology and prognosis, but it is only limitedly available in clinical practice. The aim of this investigation was to explore what additional prognostic information could be gained from detailed histomorphologic report. PATIENTS AND METHOD patients were selected retrospectively operated from 2005 to 2009 in one institution and charts were revised. Beyond age, tumour and nodal status, histologic grade and therapy, the additional pathologic characteristics were also involved in analysis: necrosis, lymphocytic infiltration, peritumoural vascular invasion (PVI), perineural invasion, DCIS extent and grade, perinodal spread, mitotic activity index (MAI). RESULTS 295 early TNBC were involved. In univariate survival analysis with a mean follow-up of 3.57 years the tumour size, the nodal status, type of operation (conservation or mastectomy), irradiation, PVI and perinodal spread proved to be significantly connected with both disease free survival (DFS) and breast cancer specific overall survival (BSOS), and necrosis and chemotherapy with BSOS. Necrosis analysed together with lymphocytic infiltrate showed greater predicting power. In multivariate analysis nodal metastasis, necrosis positive/lymphacytic infiltration negative status and lack of irradiation has significant negative impact on DFS (p = <0.0001 HR:1.98 [1.4-2.77], p = <0.017 HR:2.1 [1.1-3.8], p = <0.001 HR:0.25 [0.11-0.57], respectively) and BSOS (p = <0.0001 HR:2.47 [1.8-3.4], p = <0.017 HR:3.7 [1.6-8.2], p = <0.0017 HR:0.24 [0.1-0.58], respectively). For DFS perivascular invasion also showed significant effect (p = <0.042 HR:2.5 [1.0-6.0]). Nodal status was the strongest prognostic parameter but other histomorphologic parameters can be used for prognosis prediction.
Collapse
|
24
|
Bertucci F, Finetti P, Birnbaum D. Basal breast cancer: a complex and deadly molecular subtype. Curr Mol Med 2012; 12:96-110. [PMID: 22082486 PMCID: PMC3343384 DOI: 10.2174/156652412798376134] [Citation(s) in RCA: 152] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 10/24/2011] [Accepted: 11/02/2011] [Indexed: 12/15/2022]
Abstract
During the last decade, gene expression profiling of breast cancer has revealed the existence of five molecular subtypes and allowed the establishment of a new classification. The basal subtype, which represents 15-25% of cases, is characterized by an expression profile similar to that of myoepithelial normal mammary cells. Basal tumors are frequently assimilated to triple-negative (TN) breast cancers. They display epidemiological and clinico-pathological features distinct from other subtypes. Their pattern of relapse is characterized by frequent and early relapses and visceral locations. Despite a relative sensitivity to chemotherapy, the prognosis is poor. Recent characterization of their molecular features, such as the dysfunction of the BRCA1 pathway or the frequent expression of EGFR, provides opportunities for optimizing the systemic treatment. Several clinical trials dedicated to basal or TN tumors are testing cytotoxic agents and/or molecularly targeted therapies. This review summarizes the current state of knowledge of this aggressive and hard-to-treat subtype of breast cancer.
Collapse
Affiliation(s)
- F Bertucci
- Département d'Oncologie Médicale, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille, UMR891 Inserm, Marseille, France.
| | | | | |
Collapse
|
25
|
Vranic S, Frkovic-Grazio S, Bilalovic N, Gatalica Z. Angiogenesis in triple-negative adenoid cystic carcinomas of the breast. Virchows Arch 2011; 459:377-82. [DOI: 10.1007/s00428-011-1144-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Revised: 08/19/2011] [Accepted: 08/22/2011] [Indexed: 11/28/2022]
|